Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 295
Filter
1.
Gut Microbes ; 16(1): 2350784, 2024.
Article in English | MEDLINE | ID: mdl-38727219

ABSTRACT

The gut microbiota constitutes a vast ecological system within the human body, forming a mutually interdependent entity with the host. In recent years, advancements in molecular biology technologies have provided a clearer understanding of the role of the gut microbiota. They not only influence the local immune status and metabolic functions of the host's intestinal tract but also impact the functional transformation of hematopoietic stem cells (HSCs) through the gut-blood axis. In this review, we will discuss the role of the gut microbiota in influencing hematopoiesis. We analyze the interactions between HSCs and other cellular components, with a particular emphasis on the direct functional regulation of HSCs by the gut microbiota and their indirect influence through cellular components in the bone marrow microenvironment. Additionally, we propose potential control targets for signaling pathways triggered by the gut microbiota to regulate hematopoietic function, filling crucial knowledge gaps in the development of this research field.


Subject(s)
Gastrointestinal Microbiome , Hematopoiesis , Hematopoietic Stem Cells , Hematopoiesis/physiology , Gastrointestinal Microbiome/physiology , Humans , Hematopoietic Stem Cells/microbiology , Animals , Signal Transduction , Bacteria/metabolism , Bacteria/classification , Bacteria/genetics , Gastrointestinal Tract/microbiology , Bone Marrow/microbiology , Bone Marrow/physiology
2.
Sci Adv ; 9(36): eadf9904, 2023 09 08.
Article in English | MEDLINE | ID: mdl-37672586

ABSTRACT

Hematopoietic stem and progenitor cells (HSPCs) respond to infection by proliferating and generating in-demand neutrophils through a process called emergency granulopoiesis (EG). Recently, infection-induced changes in HSPCs have also been shown to underpin the longevity of trained immunity, where they generate innate immune cells with enhanced responses to subsequent microbial threats. Using larval zebrafish to live image neutrophils and HSPCs, we show that infection-experienced HSPCs generate neutrophils with enhanced bactericidal functions. Transcriptomic analysis of EG neutrophils uncovered a previously unknown function for mitochondrial reactive oxygen species in elevating neutrophil bactericidal activity. We also reveal that driving expression of zebrafish C/EBPß within infection-naïve HSPCs is sufficient to generate neutrophils with similarly enhanced bactericidal capacity. Our work suggests that this demand-adapted source of neutrophils contributes to trained immunity by providing enhanced protection toward subsequent infections. Manipulating demand-driven granulopoiesis may provide a therapeutic strategy to boost neutrophil function and treat infectious disease.


Subject(s)
Bacterial Infections , Hematopoietic Stem Cells , Trained Immunity , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/microbiology , Animals , Zebrafish , Larva/immunology , Larva/microbiology , Reactive Oxygen Species/metabolism , Bacterial Infections/immunology
3.
Blood ; 139(1): 44-58, 2022 01 06.
Article in English | MEDLINE | ID: mdl-34525198

ABSTRACT

Aging is associated with impaired hematopoietic and immune function caused in part by decreased fitness in the hematopoietic stem cell (HSC) population and an increased myeloid differentiation bias. The reasons for this aging-associated HSC impairment are incompletely understood. Here we demonstrate that older specific pathogen free (SPF) wild-type (WT) mice in contrast to young SPF mice produce more interleukin-1a and interleukin-1b (IL-1a/b) in steady-state bone marrow (BM), with most of the IL-1a/b being derived from myeloid BM cells. Furthermore, blood from steady-state older SPF WT mice contains higher levels of microbe-associated molecular patterns, specifically TLR4 and TLR8 ligands. In addition, BM myeloid cells from older mice produce more IL-1b in vitro, and older mice show higher and more durable IL-1a/b responses upon stimulation with lipopolysaccharide in vivo. To test whether HSC aging is driven by IL-1a/b, we evaluated HSCs from IL-1 receptor 1 (IL-1R1) knockout (KO) mice. Indeed, older HSCs from IL-1R1KO mice show significantly mitigated aging-associated inflammatory signatures. Moreover, HSCs from older IL-1R1KO and from germ-free mice maintain unbiased lymphomyeloid hematopoietic differentiation upon transplantation, thus resembling this functionality of young HSCs. Importantly, in vivo antibiotic suppression of microbiota or pharmacologic blockade of IL-1 signaling in older WT mice was similarly sufficient to reverse myeloid-biased output of their HSC populations. Collectively, our data define the microbiome/IL-1/IL-1R1 axis as a key, self-sustaining and also therapeutically partially reversible driver of HSC inflammaging.


Subject(s)
Hematopoietic Stem Cells/metabolism , Inflammation/metabolism , Interleukin-1alpha/metabolism , Interleukin-1beta/metabolism , Microbiota , Aging , Animals , Cellular Senescence , Hematopoiesis , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/microbiology , Inflammation/microbiology , Mice , Mice, Knockout
4.
Cell ; 183(3): 752-770.e22, 2020 10 29.
Article in English | MEDLINE | ID: mdl-33125891

ABSTRACT

A greater understanding of hematopoietic stem cell (HSC) regulation is required for dissecting protective versus detrimental immunity to pathogens that cause chronic infections such as Mycobacterium tuberculosis (Mtb). We have shown that systemic administration of Bacille Calmette-Guérin (BCG) or ß-glucan reprograms HSCs in the bone marrow (BM) via a type II interferon (IFN-II) or interleukin-1 (IL1) response, respectively, which confers protective trained immunity against Mtb. Here, we demonstrate that, unlike BCG or ß-glucan, Mtb reprograms HSCs via an IFN-I response that suppresses myelopoiesis and impairs development of protective trained immunity to Mtb. Mechanistically, IFN-I signaling dysregulates iron metabolism, depolarizes mitochondrial membrane potential, and induces cell death specifically in myeloid progenitors. Additionally, activation of the IFN-I/iron axis in HSCs impairs trained immunity to Mtb infection. These results identify an unanticipated immune evasion strategy of Mtb in the BM that controls the magnitude and intrinsic anti-microbial capacity of innate immunity to infection.


Subject(s)
Hematopoietic Stem Cells/microbiology , Immunity , Mycobacterium tuberculosis/physiology , Myelopoiesis , Animals , Bone Marrow Cells/metabolism , Cell Proliferation , Disease Susceptibility , Homeostasis , Interferon Type I/metabolism , Iron/metabolism , Kinetics , Lung/microbiology , Lung/pathology , Macrophages/immunology , Mice, Inbred C57BL , Myeloid Cells/metabolism , Necrosis , Signal Transduction , Transcription, Genetic , Tuberculosis/immunology , Tuberculosis/microbiology , Tuberculosis/pathology
5.
Scand J Immunol ; 92(5): e12957, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32767789

ABSTRACT

Bone marrow haematopoietic stem and progenitor cells (HSPCs) express pattern recognition receptors such as Toll-like receptors (TLRs) to sense microbial products and activation of these innate immune receptors induces cytokine expression and redirects bone marrow haematopoiesis towards the increased production of myeloid cells. Secreted cytokines by HSPCs in response to TLR ligands can act in an autocrine or paracrine manner to regulate haematopoiesis. Moreover, tonic activation of HSPCs by microbiota-derived compounds might educate HSPCs to produce superior myeloid cells equipped with innate memory responses to combat pathogens. While haematopoietic stem cell activation through TLRs meets the increased demand for blood leucocytes to protect the host against infection, persistent exposure to inflammatory cytokines or microbial products might impair their function and even induce malignant transformation. This review highlights the potential outcomes of HSPCs in response to TLR ligands.


Subject(s)
Bone Marrow Cells/immunology , Hematopoietic Stem Cells/immunology , Microbiota/immunology , Myeloid Cells/immunology , Receptors, Pattern Recognition/immunology , Animals , Bone Marrow Cells/metabolism , Bone Marrow Cells/microbiology , Cytokines/immunology , Cytokines/metabolism , Hematopoiesis/immunology , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/microbiology , Humans , Myeloid Cells/metabolism , Receptors, Pattern Recognition/metabolism , Toll-Like Receptors/immunology , Toll-Like Receptors/metabolism
6.
Blood ; 136(10): 1144-1154, 2020 09 03.
Article in English | MEDLINE | ID: mdl-32438398

ABSTRACT

Hematopoiesis is a system that provides red blood cells (RBCs), leukocytes, and platelets, which are essential for oxygen transport, biodefense, and hemostasis; its balance thus affects the outcome of various disorders. Here, we report that stem cell antigen-1 (Sca-1), a cell surface marker commonly used for the identification of multipotent hematopoietic progenitors (Lin-Sca-1+c-Kit+ cells; LSKs), is not suitable for the analysis of hematopoietic responses under biological stresses with interferon production. Lin-Sca-1-c-Kit+ cells (LKs), downstream progenitors of LSKs, acquire Sca-1 expression upon inflammation, which makes it impossible to distinguish between LSKs and LKs. As an alternative and stable marker even under such stresses, we identified CD86 by screening 180 surface markers. The analysis of infection/inflammation-triggered hematopoiesis on the basis of CD86 expression newly revealed urgent erythropoiesis producing stress-resistant RBCs and intact reconstitution capacity of LSKs, which could not be detected by conventional Sca-1-based analysis.


Subject(s)
B7-2 Antigen/metabolism , Bacterial Infections/complications , Cell Differentiation , Hematopoiesis , Hematopoietic Stem Cells/pathology , Inflammation/complications , Animals , Antigens, Ly/metabolism , Bacteria/metabolism , Cells, Cultured , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/microbiology , Lipopolysaccharides , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Proto-Oncogene Proteins c-kit/metabolism
7.
Front Immunol ; 11: 123, 2020.
Article in English | MEDLINE | ID: mdl-32117273

ABSTRACT

Over the last century, the alarming surge in allergy and autoimmune disease has led to the hypothesis that decreasing exposure to microbes, which has accompanied industrialization and modern life in the Western world, has fundamentally altered the immune response. In its current iteration, the "hygiene hypothesis" suggests that reduced microbial exposures during early life restricts the production and differentiation of immune cells suited for immune regulation. Although it is now well-appreciated that the increase in hypersensitivity disorders represents a "perfect storm" of many contributing factors, we argue here that two important considerations have rarely been explored. First, the window of microbial exposure that impacts immune development is not limited to early childhood, but likely extends into the womb. Second, restricted microbial interactions by an expectant mother will bias the fetal immune system toward hypersensitivity. Here, we extend this discussion to hypothesize that the cell types sensing microbial exposures include fetal hematopoietic stem cells, which drive long-lasting changes to immunity.


Subject(s)
Fetus/immunology , Hygiene Hypothesis , Hypersensitivity/immunology , Immune System/immunology , Adult , Child , Female , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/microbiology , Host Microbial Interactions/immunology , Humans , Infant, Newborn , Inflammation/immunology , Microbial Interactions/immunology , Pregnancy , T-Lymphocytes, Helper-Inducer/immunology
8.
J Infect Dis ; 219(5): 685-694, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30376080

ABSTRACT

Latent tuberculosis has been recognized for over a century, but discovery of new niches, where Mycobacterium tuberculosis resides, continues. We evaluated literature on M.tuberculosis locations during latency, highlighting that mesenchymal and hematopoietic stem cells harbor organisms in sensitized asymptomatic individuals.


Subject(s)
Hematopoietic Stem Cells/microbiology , Latent Tuberculosis/microbiology , Latent Tuberculosis/pathology , Mesenchymal Stem Cells/microbiology , Mycobacterium tuberculosis/isolation & purification , Phagocytes/microbiology , Humans , Mycobacterium tuberculosis/growth & development
9.
J Infect Dis ; 217(10): 1667-1671, 2018 04 23.
Article in English | MEDLINE | ID: mdl-29471332

ABSTRACT

Persistence of Mycobacterium tuberculosis within human bone marrow stem cells has been identified as a potential bacterial niche during latent tuberculosis. Using a murine model of tuberculosis, we show here that bone marrow stem and progenitor cells containing M. tuberculosis propagated tuberculosis when transferred to naive mice, given that both transferred cells and recipient mice were unable to express inducible nitric oxide synthase, which mediates killing of intracellular bacteria via nitric oxide. Our findings suggest that bone marrow stem and progenitor cells containing M. tuberculosis propagate hallmarks of disease if nitric oxide-mediated killing of bacteria is defective.


Subject(s)
Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/microbiology , Mycobacterium tuberculosis/pathogenicity , Nitric Oxide Synthase Type II/metabolism , Stem Cells/metabolism , Stem Cells/microbiology , Tuberculosis/metabolism , Animals , Disease Models, Animal , Hematopoietic Stem Cell Transplantation/methods , Mice , Mice, Inbred C57BL , Nitric Oxide/metabolism , Tuberculosis/microbiology
10.
PLoS One ; 12(1): e0169119, 2017.
Article in English | MEDLINE | ID: mdl-28046053

ABSTRACT

An estimated third of the world's population is latently infected with Mycobacterium tuberculosis (Mtb), with no clinical signs of tuberculosis (TB), but lifelong risk of reactivation to active disease. The niches of persisting bacteria during latent TB infection remain unclear. We detect Mtb DNA in peripheral blood selectively in long-term repopulating pluripotent hematopoietic stem cells (LT-pHSCs) as well as in mesenchymal stem cells from latently infected human donors. In mice infected with low numbers of Mtb, that do not develop active disease we, again, find LT-pHSCs selectively infected with Mtb. In human and mouse LT-pHSCs Mtb are stressed or dormant, non-replicating bacteria. Intratracheal injection of Mtb-infected human and mouse LT-pHSCs into immune-deficient mice resuscitates Mtb to replicating bacteria within the lung, accompanied by signs of active infection. We conclude that LT-pHSCs, together with MSCs of Mtb-infected humans and mice serve as a hitherto unappreciated quiescent cellular depot for Mtb during latent TB infection.


Subject(s)
Hematopoietic Stem Cells/microbiology , Latent Tuberculosis/microbiology , Mesenchymal Stem Cells/microbiology , Mycobacterium tuberculosis , Adult , Animals , Antigens, CD34/metabolism , Bone Marrow Cells/metabolism , Cell Separation , Female , Flow Cytometry , Humans , Lung/microbiology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Signaling Lymphocytic Activation Molecule Family Member 1/metabolism , Young Adult
11.
Sci Rep ; 6: 36853, 2016 11 11.
Article in English | MEDLINE | ID: mdl-27833150

ABSTRACT

Emergency granulopoiesis occurs in response to severe microbial infection. However, whether and how other blood components, particularly monocytes/macrophages and their progenitors, including hematopoietic stem/progenitor cells (HSPCs), participate in the process and the underlying molecular mechanisms remain unknown. In this study, we challenged zebrafish larvae via direct injection of Escherichia coli into the bloodstream, which resulted in systemic inoculation with this microbe. The reaction of hematopoietic cells, including HSPCs, in the caudal hematopoietic tissue was carefully analysed. Both macrophages and neutrophils clearly expanded following the challenge. Thus, emergency myelopoiesis, including monopoiesis and granulopoiesis, occurred following systemic bacterial infection. The HSPC reaction was dependent on the bacterial burden, manifesting as a slight increase under low burden, but an obvious reduction following the administration of an excessive volume of bacteria. Pu.1 was important for the effective elimination of the microbes to prevent excessive HSPC apoptosis in response to stress. Moreover, Pu.1 played different roles in steady and emergency monopoiesis. Although Pu.1 was essential for normal macrophage development, it played suppressive roles in emergency monopoiesis. Overall, our study established a systemic bacterial infection model that led to emergency myelopoiesis, thereby improving our understanding of the function of Pu.1 in this scenario.


Subject(s)
Escherichia coli Infections/pathology , Escherichia coli/immunology , Myelopoiesis , Animals , Apoptosis , Cell Proliferation , Escherichia coli Infections/immunology , Escherichia coli Infections/microbiology , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/microbiology , Larva/microbiology , Macrophages/immunology , Macrophages/microbiology , Neutrophils/immunology , Neutrophils/microbiology , Phagocytosis , Zebrafish
12.
Sci Rep ; 6: 28082, 2016 06 16.
Article in English | MEDLINE | ID: mdl-27306736

ABSTRACT

Haematopoietic stem/progenitor cells (HSPCs) can mobilise into blood and produce immune cell lineages following stress. However, the homeostasis and function of HSPCs after infection in teleosts are less well known. Here, we report that Listonella anguillarum infection enhances HSPC mobilisation and reduces their differentiation into myeloid cells in ayu (Plecoglossus altivelis), an aquacultured teleost in East Asia. We established a colony-forming unit culture (CFU-C) assay to measure HSPCs using conditioned medium from peripheral blood mononuclear cells stimulated with phytohaemagglutinin. The number of CFU-Cs decreased in the head kidney and increased in the blood and spleen of ayu infected with L. anguillarum. HSPC mobilisation after L. anguillarum infection was mediated by norepinephrine. Furthermore, HSPCs from ayu treated with L. anguillarum lipopolysaccharide (LPS) showed defective myeloid differentiation and could no longer rescue L. anguillarum-infected ayu. HSPC expansion was suppressed after L. anguillarum infection or its LPS treatment in vitro. These results reveal a link between HSPC regulation and pathogen infection in teleosts.


Subject(s)
Fish Diseases/pathology , Gram-Negative Bacterial Infections/veterinary , Hematopoietic Stem Cells/pathology , Leukocytes, Mononuclear/pathology , Listonella/pathogenicity , Osmeriformes/microbiology , Animals , Cells, Cultured , Fish Diseases/microbiology , Gram-Negative Bacterial Infections/microbiology , Hematopoietic Stem Cells/microbiology , Leukocytes, Mononuclear/microbiology
13.
Transfus Apher Sci ; 55(1): 125-8, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27184293

ABSTRACT

INTRODUCTION: Microbial contamination can be a marker for faulty process and is assumed to play an important role in the collection of hematopoietic progenitor cell (HPC) and infusion procedure. We aimed to determine the microbial contamination rates and evaluate the success of hematopoietic cell transplantation (HCT) in patients who received contaminated products. PATIENTS-METHODS: We analyzed microbial contamination records of HPC grafts between 2012 and 2015, retrospectively. Contamination rates of autologous donors were evaluated for at three steps: at the end of mobilization, following processing with dimethyl sulfoxide, and just before stem cell infusion. Grafts of allogeneic donors were assessed only before HCT. RESULT: A total of 445 mobilization procedures were carried out on 333 (167 autologous and 166 allogeneic) donors. The microbiological contamination of peripheral blood (323/333 donations) and bone marrow (10/333 donations) products were analyzed. Bacterial contamination was detected in 18 of 1552 (1.15 %) culture bottles of 333 donors. During the study period 248 patients underwent HCT and among these patients microbial contamination rate on sample basis was 1.3 % (16/1212). Microbial contamination detected in nine patients (7 autologous; 2 allogeneic). In 8 of 9 patients, a febrile neutropenic attack was observed. The median day for the neutropenic fever was 4 days (0-9). None of the patients died within the post-transplant 30 days who received contaminated products. CONCLUSION: The use of contaminated products with antibiotic prophylaxis may be safe in terms of the first day of fever, duration of fever, neutrophil, platelet engraftment and duration of hospitalization.


Subject(s)
Hematologic Neoplasms/microbiology , Hematologic Neoplasms/therapy , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/microbiology , Adolescent , Adult , Aged , Allografts , Autografts , Female , Humans , Male , Middle Aged
14.
Inflamm Bowel Dis ; 22(4): 763-73, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26963567

ABSTRACT

BACKGROUND: NOD2 mutations are associated with Crohn's disease (CD). Both CD (in human) and Nod2 deficiency (in mice) are characterized by increased mucosal CD4 T-cells, an altered permeability and a microbial dysbiosis. However, the respective roles of the gut epithelial and immune compartments on the phenotype are not known. METHODS: Microbial composition, epithelial peptide secretion, intestinal permeability, and immune cell composition of Peyer patches were studied in Nod2 knock-out mice transplanted with wild-type bone marrow cells and vice versa. RESULTS: The nonhematopoietic cells control the microbiota composition and epithelial secretion of mucins and antimicrobial peptides. These parameters are correlated with recurrent associations between bacterial species and luminal products. In contrast, Nod2 in the hematopoietic compartment regulates the epithelial permeability and the gut-associated lymphoid tissue independently of the bacterial composition. CONCLUSIONS: The immune system and the gut permeability in one hand and the microbial and epithelial peptide compositions in the other hand are separate couples of interdependent parameters, both controlled by Nod2 in either the hematopoietic or nonhematopoietic lineages.


Subject(s)
Dysbiosis/microbiology , Gastrointestinal Microbiome , Hematopoietic Stem Cells/microbiology , Homeostasis/physiology , Intestinal Mucosa/microbiology , Nod2 Signaling Adaptor Protein/physiology , Animals , Dysbiosis/metabolism , Dysbiosis/pathology , Hematopoietic Stem Cells/metabolism , Intestinal Mucosa/metabolism , Mice , Mice, Knockout
15.
Cell Microbiol ; 18(10): 1339-48, 2016 10.
Article in English | MEDLINE | ID: mdl-26990252

ABSTRACT

Haemolytic anaemia is one of the characteristics of life-threatening extraintestinal complications in humans during infection with enterohaemorrhagic Escherichia coli (EHEC). Shiga toxins (Stxs) of EHEC preferentially damage microvascular endothelial cells of the kidney and the brain, whereby occluded small blood vessels may elicit anaemia through mechanical erythrocyte disruption. Here we show for the first time that Stx2a, the major virulence factor of EHEC, is also capable of direct targeting developing human erythrocytes. We employed an ex vivo erythropoiesis model using mobilized CD34(+) haematopoietic stem/progenitor cells from human blood and monitored expression of Stx receptors and Stx2a-mediated cellular injury of developing erythrocytes. CD34(+) haematopoietic stem/progenitor cells were negative for Stx2a receptors and resistant towards the toxin. Expression of Stx2a-binding glycosphingolipids and toxin sensitivity was apparent immediately after initiation of erythropoietic differentiation, peaked for basophilic and polychromatic erythroblast stages and declined during maturation into orthochromatic erythroblasts and reticulocytes, which became highly refractory to Stx2a. The observed Stx-mediated toxicity towards erythroblasts during the course of erythropoiesis might contribute, although speculative at this stage of research, to the anaemia caused by Stx-producing pathogens.


Subject(s)
Enterohemorrhagic Escherichia coli/physiology , Hematopoietic Stem Cells/physiology , Shiga Toxin/pharmacology , Cell Survival , Cells, Cultured , Erythrocytes/microbiology , Erythrocytes/physiology , Hematopoiesis/immunology , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/microbiology , Humans
16.
Cell Rep ; 11(1): 71-84, 2015 Apr 07.
Article in English | MEDLINE | ID: mdl-25843711

ABSTRACT

Upon systemic bacterial infection, hematopoietic stem and progenitor cells (HSPCs) migrate to the periphery in order to supply a sufficient number of immune cells. Although pathogen-associated molecular patterns reportedly mediate HSPC activation, how HSPCs detect pathogen invasion in vivo remains elusive. Bacteria use the second messenger bis-(3'-5')-cyclic dimeric guanosine monophosphate (c-di-GMP) for a variety of activities. Here, we report that c-di-GMP comprehensively regulated both HSPCs and their niche cells through an innate immune sensor, STING, thereby inducing entry into the cell cycle and mobilization of HSPCs while decreasing the number and repopulation capacity of long-term hematopoietic stem cells. Furthermore, we show that type I interferon acted as a downstream target of c-di-GMP to inhibit HSPC expansion in the spleen, while transforming growth factor-ß was required for c-di-GMP-dependent splenic HSPC expansion. Our results define machinery underlying the dynamic regulation of HSPCs and their niches during bacterial infection through c-di-GMP/STING signaling.


Subject(s)
Cyclic GMP/analogs & derivatives , Hematopoietic Stem Cells/metabolism , Immunity, Innate , Interferon Regulatory Factor-3/genetics , Membrane Proteins/genetics , Animals , Bacteria/metabolism , Bacteria/pathogenicity , Cyclic GMP/administration & dosage , Cyclic GMP/immunology , Cyclic GMP/metabolism , Gene Expression Regulation , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/microbiology , Interferon Regulatory Factor-3/immunology , Membrane Proteins/immunology , Mice , Signal Transduction/drug effects , Signal Transduction/immunology , Stem Cell Niche/immunology , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/immunology
17.
Inflamm Bowel Dis ; 21(3): 543-55, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25659087

ABSTRACT

BACKGROUND: Ileal Crohn's disease is related to NOD2 mutations and to a gut barrier dysfunction. Pseudomonas fluorescens has also been associated with ileal Crohn's disease. The aim of this study was to determine the impact of P. fluorescens on the paracellular permeability in ileum and Peyer's patches. METHODS: To explore this question, in vivo and ex vivo experiments were performed in wild-type, Nod2, Nod2, and IL-1R mice together with in vitro analyses using the Caco-2 (epithelial) and the THP-1 (monocyte) human cell lines. RESULTS: Pseudomonas fluorescens increased the paracellular permeability of the intestinal mucosa through the secretion of IL-1ß by the immune cell populations and the activation of myosin light chain kinase in the epithelial cells. Induction of the IL-1ß pathway required the expression of Nod2 in the hematopoietic compartment, and muramyl dipeptide (a Nod2 ligand) had an inhibitory effect. CONCLUSIONS: Pseudomonas fluorescens thus alters the homeostasis of the epithelial barrier function by a mechanism similar to that previously observed for Yersinia pseudotuberculosis. This work further documents a putative role of psychrotrophic bacteria in Crohn's disease.


Subject(s)
Hematopoietic Stem Cells/metabolism , Intestinal Mucosa/metabolism , Macrophages/metabolism , Nod2 Signaling Adaptor Protein/physiology , Pseudomonas fluorescens/physiology , Receptors, Interleukin-1/physiology , Animals , Blotting, Western , Caco-2 Cells , Cell Membrane Permeability , Cells, Cultured , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/microbiology , Humans , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Intestinal Mucosa/cytology , Intestinal Mucosa/microbiology , Macrophages/cytology , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nod2 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
18.
Vox Sang ; 108(3): 314-7, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25471358

ABSTRACT

In order to generate standardized conditions for the microbiological control of HPCs, the PEI recommended defined steps for validation that will lead to extensive validation as shown in this study, where a possible validation principle for the microbiological control of allogeneic SCPs is presented. Although it could be demonstrated that automated culture improves microbial safety of cellular products, the requirement for extensive validation studies needs to be considered.


Subject(s)
Cell Culture Techniques/standards , Guidelines as Topic , Hematopoietic Stem Cells/microbiology , Cell Culture Techniques/methods , Cells, Cultured , Germany , Humans
19.
Transplant Proc ; 46(8): 2873-6, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25380939

ABSTRACT

Hematopoietic stem cells (HSC) derived from peripheral blood (PB) and bone marrow (BM) are frequently used for autologous and allogenic transplantations. Establishing quality control at appropriate steps of the stem cell preparation process is crucial for a successful transplantation. Microbial contamination of haematopoietic stem cells is rare but could cause a potentially mortal complication of a stem cells transplantation. We investigated the microbiological contamination of PB (291 donations) and BM (39 donations) products. Microbial cultures of 330 donations between January 2012 and June 2013 were retrospectively analyzed after the collection and preparation steps. The microbiological analysis was performed with an automated system. Hematopoietic stem cells were processed in a closed system. Additionally, in this report the environment of the working areas of stem cell preparation was monitored. We analyzed microbial contamination of the air in a class I laminar air flow clean bench at the time of preparation and in the laboratory once per month. We reported 9 (2.73%) contaminated HSC products. The most frequent bacteria isolated from PB and BM products were Bacillus species. Coagulase-negative staphylococci and Micrococcus species were the most frequent micro-organisms detected in the air microbial control. Microbial control results are necessary for the safety of hematopoietic stem cell products transplantation. Microbial control of hematopoietic stem cell products enables an early contamination detection and allows for knowledgeable decision making concerning either discarding the contaminated product or introducing an efficient antibiotic therapy. Each step of cell processing may cause a bacterial contamination. A minimum of manipulation steps is crucial for increasing the microbial purity of the transplant material. Also, the air contamination control is essential to ensure the highest quality standards of HSC products preparation.


Subject(s)
Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/microbiology , Bacillus/isolation & purification , Bone Marrow , Bone Marrow Cells/microbiology , Humans , Micrococcus/isolation & purification , Retrospective Studies , Staphylococcus/isolation & purification
20.
Int Immunopharmacol ; 22(1): 209-21, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24975836

ABSTRACT

This work evaluated the capacity of two probiotic strains, Lactobacillus casei CRL431 and Lactobacillus rhamnosus CRL1506, to protect against myelosuppression and immunosuppression in cyclophosphamide (Cy)-treated mice. Changes in mature granulocytes and progenitor cells in bone marrow (BM) and blood were studied. In addition, the ability of probiotics to accelerate the recovery of the immune response against the opportunistic pathogen Candida albicans was evaluated. We demonstrated for the first time that the preventive treatment with immunomodulatory lactobacilli such as L. casei CRL431 or L. rhamnosus CRL1506 was able to increase immature myeloid progenitors in the BM, allowing an early recovery of myeloid cells after Cy administration. Probiotic lactobacilli were also capable to induce an early recovery of neutrophils in blood, improve phagocytic cells recruitment to infectious sites and increase the resistance against the opportunistic pathogen C. albicans. Although deeper studies regarding the cellular and molecular mechanisms of probiotic actions are needed, these findings support the idea that strains like CRL431 and CRL1506 may accelerate the recovery of Cy-caused immunosuppression by immunopotentiating myeloid cells. Then, probiotic lactobacilli have the potential to be used as alternatives for lessening chemotherapy-induced immunosuppression in cancer patients.


Subject(s)
Candida albicans/immunology , Candidiasis/immunology , Granulocytes/immunology , Hematopoietic Stem Cells/immunology , Lacticaseibacillus casei/immunology , Lacticaseibacillus rhamnosus/immunology , Myeloid Progenitor Cells/immunology , Animals , Candidiasis/microbiology , Cell Movement , Cells, Cultured , Cyclophosphamide/metabolism , Granulocytes/microbiology , Hematopoietic Stem Cells/microbiology , Immunity, Innate , Immunomodulation , Immunosuppression Therapy , Male , Mice , Myeloid Progenitor Cells/microbiology , Neutrophils/immunology , Neutrophils/microbiology , Phagocytes/immunology , Phagocytes/microbiology , Probiotics/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...